EP2101571B1 - Ornithine conjugates of amphetamine and processes for making and using the same - Google Patents

Ornithine conjugates of amphetamine and processes for making and using the same Download PDF

Info

Publication number
EP2101571B1
EP2101571B1 EP07869098A EP07869098A EP2101571B1 EP 2101571 B1 EP2101571 B1 EP 2101571B1 EP 07869098 A EP07869098 A EP 07869098A EP 07869098 A EP07869098 A EP 07869098A EP 2101571 B1 EP2101571 B1 EP 2101571B1
Authority
EP
European Patent Office
Prior art keywords
amphetamine
conjugate
composition
salt
ornithine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP07869098A
Other languages
German (de)
French (fr)
Other versions
EP2101571A1 (en
EP2101571A4 (en
Inventor
Travis C. Mickle
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zevra Therapeutics Inc
Original Assignee
KemPharm Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by KemPharm Inc filed Critical KemPharm Inc
Publication of EP2101571A1 publication Critical patent/EP2101571A1/en
Publication of EP2101571A4 publication Critical patent/EP2101571A4/en
Application granted granted Critical
Publication of EP2101571B1 publication Critical patent/EP2101571B1/en
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • A61K31/198Alpha-aminoacids, e.g. alanine, edetic acids [EDTA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents

Definitions

  • compositions comprising at least one amphetamine conjugated with the non-standard amino acid ornithine, or a salt thereof, which can diminish or eliminate pharmacological activity of the amphetamine until released in vivo.
  • the non-standard amino acid conjugate(s) is amphetamine in a prodrug form, and can be converted into its active form in the body by normal metabolic processes.
  • ornithine conjugates of amphetamine are believed to be safer than other sustained release forms of amphetamine by providing controlled blood levels for a prolonged period of time, thus preventing the rebound effect, cardiovascular stress and euphoria associated with conventional stimulant treatment options.
  • compositions comprising amphetamine prodrugs of the present technology are resistant to abuse by parenteral routes of administration, such as intravenous "shooting,” intranasal “snorting,” or inhalation “smoking,” that are often employed during illicit use.
  • the present technology thus provides a stimulant based treatment modality and dosage form for certain disorders requiring the stimulation of the CNS such as ADHD, ADD, obesity, narcolepsy, appetite suppressant, depression, anxiety, and wakefulness with reduced or prevented abuse potential.
  • carbodiimides One category of acid activating agent well known in the art is carbodiimides.
  • carbodiimide acid activating agents include, but are not limited to, dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide (EDCI), and diisopropylcarbodiimide (DIPCDI).
  • DCC dicyclohexylcarbodiimide
  • EDCI 1-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide
  • DIPCDI diisopropylcarbodiimide
  • the N-protected amino acid conjugate of amphetamine resulting from the reaction of the N-protected amino acid and amphetamine can then be de- or un-protected with a strong acid to produce the corresponding final salt form of the non-standard amino acid conjugate of amphetamine.
  • Scheme 1 below shows
  • Film-coated tablets may be prepared by coating tablets using techniques such as rotating pan coating methods or air suspension methods to deposit a contiguous film layer on a tablet.
  • the amphetamine composition used may be adapted for oral administration, and the amphetamine prodrug may be resistant to release amphetamine from the ornithine moiety when the composition is administered parenterally, such as intranasally or intravenously.
  • amphetamine may be released from the ornithine moiety in the presence of the intestinal tract.
  • the amphetamine composition used may be in the form of a tablet, capsule, oral film, oral solution, oral suspension, or other oral dosage form discussed herein.

Abstract

Disclosed are polar, hydrophilic stimulant prodrug compositions comprising at least one stimulant chemically attached to a polar hydrophilic ligand, a salt thereof, a derivative thereof, or a combination thereof. Also disclosed are non-standard amino acid conjugates of amphetamine. Methods of making and using the same are also disclosed.

Description

    BACKGROUND OF THE INVENTION
  • The present technology describes, in general, novel prodrugs/compositions of the stimulant amphetamine (i.e., 1-pbenylpropan-2-amine) and, in particular, conjugates of amphetamine with the non-standard amino acid ornithine salts thereof, derivatives thereof, or combinations thereof. Additionally, the presently described technology also relates generally to uses of these new prodrugs/compositions.
  • The presently described technology in at least one aspect is focused on a slow/sustained controlled release composition of amphetamine, in prodrug form, that allows slow/sustained/controlled delivery of the stimulant into the blood system of a human or animal within a safe therapeutic window upon oral administration. At least some compositions/formulations of the current technology can lessen the rebound effect, cardiovascular stress, addiction/abuse potential and/or other common stimulant side effects associated with amphetamine and similar compounds. Such compositions may also increase the duration of therapeutic efficacy, ease of application, patient compliance and/or any combination of these characteristics when administered, in particular, orally.
  • Stimulants, including amphetamine and its derivatives, enhance the activity of the sympathetic nervous system and/or central nervous system (CNS) and are prescribed for the treatment of a range of conditions and disorders predominantly encompassing, for example, attention deficit hyperactivity disorder (ADHD), attention deficit disorder (ADD), obesity, narcolepsy, appetite suppression, depression, anxiety and wakefulness.
  • Attention deficit hyperactivity disorder (ADHD) in children has been treated with stimulants for many years. However, more recently, the increase in number of prescriptions for ADHD therapy in adult population has, at times, outperformed the growth of the pediatric market. Although there are various drugs currently in use for the treatment of ADHD, such as methylphenidate (commercially available from, for example, Novartis International AG (located in Basel, Switzerland) under the trademark Ritalin®) and non-stimulant atomoxetine (commercially from Eli Lilly and Company (located in Indianapolis, IN) as Strattera®), amphetamine has been the forerunner in ADHD therapy. Moreover, during classroom trials non-stimulants have shown to be less effective in improving behavior and attention of ADHD afflicted children than amphetamine derivatives.
  • Initial drug therapy for ADHD was limited to fast acting immediate release formulations of stimulants (e.g., Dexedrine®, pure dextroamphetamine sulfate, commercially available from Smith Kline and French located in the United Kingdom) which triggered an array of potentially undesirable side effects including, for example, fast wear-off of the therapeutic effect of the stimulant active ingredient causing rebound symptoms, cardiovascular stress/disorders (e.g., increased heart rate, hypertension, cardiomyopathy), other side effects (e.g., insomnia, euphoria, psychotic episodes), addiction and abuse.
  • Behavioral deterioration (rebound/"crashing") is observed in a significant portion of children with ADHD as the medication wears off, typically in the afternoon or early evening. Rebound symptoms include, for example, irritability, crankiness, hyperactivity worse than in the unmedicated state, sadness, crying and in rare cases psychotic episodes. The symptoms may subside quickly or last several hours. Some patients may experience rebound/crashing so severe that treatment must be discontinued. Rebound/crashing effects can also give rise to addictive behavior by enticing patients to administer additional doses of stimulant with the intent to prevent anticipated rebound/crashing negative outcomes and side effects.
  • Stimulants, such as methylphenidate and amphetamine, have shown to exhibit noradrenergic and dopaminergic effects that can lead to cardiovascular events comprising, for example, increased heart rate, hypertension, palpitations, tachycardia and in isolated cases cardiomyopathy, stroke, myocardial infarction and sudden death. Consequently, currently available stimulants expose patients with pre-existing structural cardiac abnormalities or other severe cardiac indications to even greater health risks and are frequently not used or used with caution in this population. It is notable, however, that the cardiovascular effects of stimulants, for example on heart rate and blood pressure, is dependent on the administered dose. As a result, a treatment which maintains the lowest effective stimulant blood concentrations for a therapeutically beneficial duration is believed to demonstrate fewer cardiovascular risks.
  • Amphetamine and many of its derivatives (e.g., methamphetamine, 3,4-methylenedioxy-methamphetamine/"Ecstacy") are widely abused for various purposes such as euphoria, extended periods of alertness/wakefulness, or rapid weight loss or by actual ADHD patients who developed excessive self-dosing habits to prevent rebound symptoms from manifesting, for example, in anxiety or depression. The effects desired by potential abusers originated from the stimulation of the central nervous system and prompted a Schedule II or even Schedule I classification for amphetamine (d- and 1-amphetamine individually and any combination of both are Schedule II) and certain derivatives thereof after passage of the Controlled Substance Act (CSA) in 1970. Both classifications are defined by the high propensity for abuse. Schedule II drugs have an accepted medical use while Schedule I substances do not pursuant to the CSA. So far, all amphetamine products, including compositions with sustained release formulations and prodrugs thereof, are obligated to include a black box warning on the drug label to inform patients about the potential for amphetamine abuse and dependence.
  • It has been shown in the conventional art that most side effects of amphetamines are caused by a large initial spike in blood concentration of the stimulant which quickly erodes to levels below therapeutic effectiveness (typically within 4-6 hours). As a consequence, the high potency of dextroamphetamine (d-amphetamine) was subsequently modulated by a series of new drugs with increasingly sustained release profiles achieved by delivering amphetamine more slowly into the blood stream with the goal to create safer and less abusable treatment outcomes and regimens. The methods and technologies for generating smaller spikes in drug blood concentrations include, for example, use of mixed salts and isomer compositions (i.e., different salts of d- and less potent 1-amphetamine), extended/controlled/sustained release formulations (e.g., Adderall X® commercially available from Shire U.S., Inc. located in Wayne, PA) and, most recently, prodrugs of amphetamine (Vyvanse™ also commercially available from Shire). The ideal drug treatment option should produce stimulant blood concentrations within a narrow therapeutic window for an extended time duration followed by a prolonged fade-out period in order to minimize cardiovascular stress and behavioral deterioration, and would also exhibit anti-abuse properties.
  • Besides immediate release formulations, newer sustained release formulations have been developed with the objective to provide a therapeutic treatment option that offers the convenience of a single daily dosing regimen versus multiple quotidian administrations. Such formulations also have the objective of imparting or rendering a euphoric response. Sustained release formulations commonly consist of drug particles coated with a polymer or polymer blend that delays and extends the absorption of the active drug substance by the gastrointestinal tract for a relatively defined period of time. Such formulations frequently embed the therapeutic agent/active ingredient/drug within a hydrophilic hydrocolloid gelling polymer matrix (e.g., hydroxypropyl methylcellulose, hydroxypropyl cellulose or pullulan). This dosage formulation in turn becomes a gel upon entering an acidic medium, as found in the stomach of humans and animals, thereupon slowly effusing the therapeutic agent/active ingredient/drug. However, the dosage formulation dissolves in an alkaline medium, as found in the intestines of humans and animals, concurrently liberating the drug more quickly in an uncontrolled manner. Some formulations, such as acrylic resins, acrylic latex dispersions, cellulose acetate phthalate, and hydroxypropyl methylcellulose phthalate, offer improved sustained release in the intestines by being resistant to acidic environments and dispensing the active ingredient only at elevated pH via a diffusion-erosion mechanism, either by themselves or mixed with hydrophilic polymers.
  • Sustained release formulations have been moderately effective in providing an improved and extended dosage form over immediate release tablets. Nonetheless, such formulations are potentially subject to inconsistent, erratic or premature release of the therapeutic agent due to failure of the polymer material and they also usually allow easy extraction of the active ingredient utilizing a simple physical procedure. Since single daily dose formulations contain a greater amount of amphetamine than immediate release formulations, they are more attractive to potential abusers, consequently making the extractability of drug substance an additional undesirable property. It is also, at least in part, a reason for increased drug diversion, especially evident by selling or trading of medication by school children who are ADHD patients and in possession of sustained release amphetamine capsules. The obtained stimulants are then abused by classmates without the disorder by either ingesting high doses or snorting the drug material after crushing it.
  • U.S. Pat. No. 7,105,486 (to assignee New River Pharmaceuticals, hereinafter the "'486 patent") appears to describe compounds comprising a chemical moiety (namely L-lysine) covalently attached to amphetamine, compositions thereof, and methods of using the same. Allegedly, these compounds and their compositions are useful for reducing or preventing abuse and overdose of amphetamine. The '486 patent also describes that using any amino acid other than 1-lysine (Table 46) will not give rise to the same in vivo properties demonstrated by 1-lysine-d-amphetamine (Lys-Amp, Vyvanse™). Additionally, since lysine is a natural and standard amino acid, the breakdown of the new prodrug occurs faster than desired to reduce the side effect profile. Thus, quick release of amphetamine from such standard amino acid conjugate compositions may cause an increase in blood pressure and heart rate found in other conventional stimulant treatments. As a result, there still exists a need within the art for a safer dosage form of amphetamine, and treatment regimen that is therapeutically effective and can provide sustained release and sustained therapeutic effect
  • BRIEF SUMMARY OF THE INVENTION
  • The presently described technology provides, in part, compositions comprising at least one amphetamine conjugated with the non-standard amino acid ornithine, or a salt thereof, which can diminish or eliminate pharmacological activity of the amphetamine until released in vivo. The non-standard amino acid conjugate(s) is amphetamine in a prodrug form, and can be converted into its active form in the body by normal metabolic processes. Although not wanting to be bound by any particular theory, ornithine conjugates of amphetamine are believed to be safer than other sustained release forms of amphetamine by providing controlled blood levels for a prolonged period of time, thus preventing the rebound effect, cardiovascular stress and euphoria associated with conventional stimulant treatment options.
  • Release of amphetamine following oral administration of ornithine conjugates of amphetamine can occur gradually over an extended period of time thereby eliminating unintended elevations (e.g., blood level concentration spikes) of drug levels in the bloodstream of a human or animal patient. Again not wanting to be bound by any particular theory, it is also believed that such spikes in blood levels can lead to a euphoric drug "high" and cardiovascular effects like increased blood pressure and heart rate. Additionally, sustained blood levels are achieved within an effective therapeutic range for a longer duration than other conventional therapies, thereby preventing a rebound effect.
  • At least some compositions comprising amphetamine prodrugs of the present technology are resistant to abuse by parenteral routes of administration, such as intravenous "shooting," intranasal "snorting," or inhalation "smoking," that are often employed during illicit use. The present technology thus provides a stimulant based treatment modality and dosage form for certain disorders requiring the stimulation of the CNS such as ADHD, ADD, obesity, narcolepsy, appetite suppressant, depression, anxiety, and wakefulness with reduced or prevented abuse potential. Although not wanting to be bound by any particular theory, it is believed that the treatment of such CNS conditions as noted above with compositions of the present technology results in substantially decreased abuse liability as compared to existing stimulant treatment modalities and dosage forms.
  • At least some compositions comprising amphetamine prodrugs of the present technology can also be used for treating stimulant (***e, methamphetamine) abuse and addiction, for improving battle field alertness, and/or for combating fatigue.
  • In a first aspect, the presently described technology provides a composition for treating a human or animal patient having a disorder or condition requiring the stimulation of the central nervous system of the patient, comprising at least one conjugate of amphetamine with the non-standard amino acid ornithine, a salt thereof, a derivative thereof, or a combination thereof. Preferably, at least one conjugate of amphetamine is 1-ornithine-d-amphetamine. Preferably, the composition has a reduced or prevented pharmacological activity when administered by parenteral routes. Preferably, the salt of the conjugate of amphetamine is a mesylate, a hydrochloride salt, a sulfate, an oxalate, a triflate, a citrate, a malate, a tartrate, a phosphate, a nitrate, a benzoate, or a mixture thereof. Preferably, the composition is in the form of a tablet, a capsule, a caplet, a troche, a lozenge, an oral powder, a solution, an oral film, a thin strip, or a suspension, wherein the tablet, troche, or lozenge preferably is chewable. Preferably, at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof is present in the amount of from about 1 mg to about 500 mg, preferably from about 5 mg to about 250 mg, more preferably from about 10 mg to about 100 mg. Preferably, at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof is in an amount sufficient to provide a therapeutically bioequivalent AUC when compared to amphetamine alone, but does not provide a Cmax spike or does not provide an equivalent Cmax.
  • Preferably, the non-standard amino acid is covalently attached to amphetamine through the C-terminus of the non-standard amino acid. The N-terminus or the side chain amino group of the non-standard amino acid may be in a free and unprotected state, or in the form of a salt thereof. The non-standard amino acid moiety can be derived from a non-standard amino acid that is either a dextro- (d-) or levo- (1-) form amino acid, racemic amino acid, or a mixture thereof.
  • The compositions of the present technology preferably have no or a substantially decreased pharmacological activity when administered through injection or intranasal routes of administration. However, they remain orally bioavailable. The bioavailability can be a result of the hydrolysis of the covalent linkage following oral administration. Hydrolysis is time-dependent, thereby allowing amphetamine and other metabolites such as p-hydroxyamphetamine and p-hydroxyephedrine to become available in its active form over an extended period of time. In at least one further embodiment, release of amphetamine is diminished or eliminated when the composition of the present technology is delivered by parenteral routes.
  • For example, in one embodiment, the composition of the present technology maintains its effectiveness and abuse resistance following the crushing of the tablet, capsule or other oral dosage form utilized to deliver the therapeutic component (i.e., active ingredient/drug) due to the inherent controlled release components being a property of the composition not formulation. In contrast, conventional extended release formulations used to control the release of amphetamine are subject to release of up to the entire amphetamine content immediately following crushing. When the content of the crushed tablet is injected or snorted, the large dose of amphetamine produces the "rush" effect sought by addicts.
  • In another aspect, the presently described technology provides a use of at least one conjugate of amphetamine with ornithine, a salt thereof, a derivative thereof, or a combination thereof for the preparation of a pharmaceutical composition for treating a patient having a disorder or condition requiring the stimulation of the CNS (central nervous system) of the patient, wherein the pharmaceutical composition is to be administered orally to the patient. Preferably, at least one conjugate of amphetamine is l-omithine-d-amphetamine. Preferably, said oral administration comprises administering a tablet, a capsule, a caplet, a troche, a lozenge, an oral powder, an oral solution, an oral film, a thin strip, or an oral suspension. Preferably, the pharmaceutical composition is for administering the conjugate of amphetamine in the form of a salt, wherein the salt preferably is a mesylate, a hydrochloride salt, a sulfate, an oxalate, a triflate, a citrate, a malate, a tartrate, a phosphate, a nitrate, a benzoate, or a mixture thereof. Preferably, the pharmaceutical composition contains from about 1 mg to about 500 mg, preferably from about 5 mg to about 250 mg, more preferably from about 10 mg to about 100 mg of the at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof. Preferably, the pharmaceutical composition is for administering at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof in an amount sufficient to provide a therapeutically bioequivalent area under the curve (AUC) when compared to amphetamine alone, but does not provide a Cmax spike or does not provide an equivalent Cmax. Preferably, the disorder or condition is attention deficit hyperactivity disorder, attention deficit disorder, obesity, narcolepsy, appetite suppression, depression, anxiety, wakefulness, or a combination thereof. Preferably, the blood levels of amphetamine in the patient's body can maintain a therapeutically effect level throughout a given day, and do not lead to behavioral deterioration or the rebound effect.
  • Preferably, the blood levels of amphetamine in the patient's body are not unnecessarily elevated (i.e., blood level spikes) thus preventing additional cardiovascular stress through, for example, increased blood pressure and/or heart rate.
  • Preferably, the blood levels of amphetamine in the patient's body can maintain a therapeutically effect level, but do not result in an euphoric effect (such as that observed with abuse of amphetamines).
  • Other objects, advantages and embodiments of the invention are described below and will be obvious from this description and practice of the invention.
  • BRIEF DESCRIPTION OF SEVERAL VIEWS OF THE DRAWINGS
  • Figure 1 shows the mean plasma concentration curves (n=5) of d-amphetamine released by l-homoarginine-d-amphetamine or l-lysine-d-amphetamine in the biological oral study of Example 7.
  • Figure 2 shows the relative blood levels of d-amphetamine released from both Lys-Amp and hArg-Amp in the study described in Figure 1 and Table 2.
  • Figures 3 and 4 show the difference in blood levels obtained from the study described in Figure 2.
  • Figure 5 shows the average pharmacokinetic (PK) results of the other oral studies in Example 7 for four (4) oral studies (n=20 per vehicle) of l-homoarginine-d-amphetamine and l-lysine-d-amphetamine.
  • Figure 6 shows the mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp or Orn-Amp in the oral pharmacokinetic study of Example 8.
  • Figure 7 shows the mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp, Orn-Amp and Cit-Amp in the oral pharmacokinetic study of Example 9.
  • Figure 8 shows the mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp or Orn-Amp in the intranasal study of Example 10.
  • Figure 9 shows the mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp or Orn-Amp in the intranasal study of Example 11.
  • DETAILED DESCRIPTION OF THE INVENTION
  • As used herein, a "non-standard" amino acid refers to an amino acid that is not one of the "standard" 20 amino acids and they may be derived from either natural or synthetic sources. "Non-standard" amino acids are non-essential, and are not readily incorporated into proteins of natural origin. They are either metabolites or precursors in various metabolic pathways. With the exception of selenocysteine, there is no human genetic codon for the formation of non-standard amino acids. For example, the diamino acid lysine is a standard, essential amino acid.
  • As used herein, "amphetamine" shall mean any of the sympathomimetic phenethylamine derivatives which have central nervous system stimulant activity, including, but not limited to, amphetamine (alpha-methyl-phenethylamine), methamphetamine, p-methoxyamphetamine, methylenedioxyamphetamine, 2,5-dimethoxy-4-methylamphetamine, 2,4,5-trimethoxyamphetamine, 3,4-methylenedioxy-methamphetamine, and methylphenidate.
  • As used herein, "in a manner inconsistent with the manufacturer's instructions" or similar expression is meant to include, but is not limited to, consuming amounts greater than amounts described on the label or ordered by a licensed physician, and/or altering by any means (e.g., crushing, breaking, melting, separating, etc.) the dosage formulation such that the composition may be injected, inhaled or smoked.
  • As used herein, the phrases such as "decreased," "reduced," "diminished" or "lowered" is meant to include at least a 10% change in pharmacological activity with greater percentage changes being preferred for reduction in abuse potential and overdose potential. For instance, the change may also be greater than 25%, 35%, 45%, 55%, 65%, 75%, 85%, 95%, 96%, 97%, 98%, 99%, or increments therein
  • Amphetamine includes different forms or modified forms of sympathomimetic phenethylamine derivatives. According to the presently described technology, the non-standard amino acid
    can be either the dextro- (d-) or levo- (I-) form of the amino acid, a racemic mixture of the amino acid, or a mixture thereof.
  • One group of non-standard amino acids can be represented by the following general formula:

            NH2-CH(R)-COOH

    wherein R is a side chain of one of the non-standard amino acids. R can comprise the side chain of ornithine (-CH2CH2CH2NH2) or homoarginine (-CH2CH2CH2CH2NH-(C=NH)-NH2).
  • In accordance with the presently described technology, the non-standard amino acid ornithine is attached to amphetamine to make the non-standard amino acid conjugate of amphetamine or salts thereof. Preferably, the non-standard amino acid is covalently attached to amphetamine through the C-terminus of the amino acid. The N-terminus or the side chain amino group of the amino acid may be in a free and unprotected state, or in the form of a salt thereof. Alternatively, in some embodiments, the non-standard amino acid can be attached to amphetamine through the N-terminus. Examples of salts of non-standard amino acid conjugates of amphetamine that can be formed and administered to patients in accordance with the presently described technology include, but are not limited to, mesylate, hydrochloride, sulfate, oxalate, triflate, citrate, malate, tartrate, phosphate, nitrate, and benzoate salts, and mixtures thereof.
  • Non-standard amino acid conjugates of amphetamine can be represented by the following general formula,
    Figure imgb0001
    wherein R = the side chain of any non-standard amino acid.
  • Examples of non-standard amino acids include, but are not limited to: ornithine, homoarginine, pyrrolysine, lanthionine, 2-aminoisobutyric acid, dehydroalanine, B-alanine, homocysteine, homoserine, 2-oxoarginine, gamma-aminobutyric acid (GABA), 4-amino butanoic acid, all phosphorylated standard amino acids, all hydroxylated standard amino acids, all acetylated standard amino acids, all succinated standard amino acids, all methylated standard amino acids, LL-2,6-diaminopimelic acid, 6-aminohexanoic acid, L-2-aminoadipate 6-semialdehyde, pipecolic acid, D-threo-2,4-diaminopentanoate, 2-amino-4-oxopentanoic acid, L-erythro-3,5-diaminohexanoic acid, (S)-5-amino-3-oxohexanoic acid, N6-hydroxy-L-lysine, N6-acyl-L-lysine, L-saccharopine, 5-aminovaleric acid, N6-methyl-L-lysine, N6,N6-dimethyl-L-lysine, N6,N6,N6-trimethyl-L-lysine, 3-hydroxy-N6,N6,N6-trimethyl-L-lysine, 4-trimethylammoniobutanoic acid, 5-hydroxy-L-lysine, L-citrulline, 2-oxo-4-hydroxy-5-aminovalerate, pyrrole-2-carboxylate, L-erythro-4-hydroxyglutamic acid, trans-4-hydroxy-L-proline, 4-oxoproline, N-methylglycine (sarcosine), 3-sulfino-L-alanine, O3-acetyl-L-serine, selenomethionine, selenocysteine, Se-methylselenomethionine, Se-methylselenocysteine, selenocystathionine, selenocysteine selenate, and cystathione.
  • Some structural examples of non-standard amino acids are shown below:
    Figure imgb0002
  • The amphetamine can be in d-form, 1-form, or racemic form, or can be a mixture thereof. For example, when 1-ornithine (1-2,5-diaminopentanoic acid) is used, it can be chemically conjugated to a d- or 1-amphetamine to produce a novel prodrug of amphetamine (e.g., 2,5-diamino-N(1-phenylpropan-2-yl)pentanamide).
  • To conjugate a non-standard amino acid with amphetamine, the one or more amino groups are preferably protected before the amino acid is reacted with amphetamine. The non-standard amino acid whose amino group(s) are protected can be referred to as an N-protected amino acid. Agents and methods for protecting amino groups in a reactant are known in the art. One can either protect the amino groups prior to reaction, or use commercially available N-protected amino acids directly. Preferably, the carboxylic acid group in the N-protected amino acid is activated by an acid activating agent to help the reaction of the N-protected amino acid with amphetamine. General information about the reaction of amino acids to form peptide bonds can be found in, for example, G.C. Barett, D.T. Elmare, Amino Acids and Peptides, page 151-156, Cambridge University Press, UK (1st edition, 1998); Jones, J., Amino Acid and Peptide Synthesis, pages 25-41, Oxford University Press, UK (2nd edition, 2002), which are incorporated herein by reference in their entirety.
  • One category of acid activating agent well known in the art is carbodiimides. Examples of carbodiimide acid activating agents include, but are not limited to, dicyclohexylcarbodiimide (DCC), 1-ethyl-3-(3'-dimethylaminopropyl)-carbodiimide (EDCI), and diisopropylcarbodiimide (DIPCDI). The N-protected amino acid conjugate of amphetamine resulting from the reaction of the N-protected amino acid and amphetamine can then be de- or un-protected with a strong acid to produce the corresponding final salt form of the non-standard amino acid conjugate of amphetamine. Scheme 1 below shows a general synthetic scheme when ornithine is used as the non-standard amino acid.
    Figure imgb0003
  • In accordance with some embodiments of the presently described technology, d-amphetamine (dextroamphetamine) is preferably used and 1-ornithine is a preferred non-standard amino acid. In accordance with some other embodiments, the prodrug of d-amphetamine can be used in combination with a prodrug of 1-amphetamine or 1-amphetamine itself.
  • Synthesis of Orn-Amp (l-omithine-d-amphetamine) can be accomplished in two steps as shown in reaction Scheme 2 below. The first step is the coupling of Boc-Orn(Boc)-OH with d-amphetamine using EDCI. N-hydroxysuccinimide (NHS) can be added to form an in-situ activated ester with diisopropylethylamine (DIPEA) used as a co-base. The product can then be subjected to deprotection with methanesulfonic acid which also forms the corresponding dimesylate salt.
    Figure imgb0004
  • Besides ethyl acetate, examples of other solvents that can be used include, but are not limited to, isopropyl acetate (IPAC), acetone, and dichloromethane (DCM). A mixture of different solvents can also be used. When a polar solvent is required, water, dimethylformamide (DMF), 1,4-dioxane or dimethylsulfoxide (DMSO) can be used. Co-bases such as tertiary amines may or may not be added in the coupling reaction. Examples of suitable co-bases include 1-methylmorpholine (NMM), triethylamine (TEA), etc.
  • It is important to note that preparation of Orn-Amp requires additional experimentation compared to the prior art. It has been surprisingly found that significant changes of the solubility in starting material of Boc-Orn(Boc)-OH required the use of DMF instead of less polar solvents stated previously. In addition, due to the unique solubility differences of Orn-Amp 2MsOH as compared to either Lys-Amp 2HCl or Lys-Amp 2MsOH, the procedures of the conventional art would not give rise to Orn-Amp 2MsOH without significant experimentation. Also, formation of the free base of amphetamine was performed in situ and was not isolated. The formation of the activated ester was performed in situ with the addition reaction following in the same reaction vessel. Quite surprisingly, these changes to solubility and reaction conditions are not readily apparent from previously published procedures or from the overall structures themselves and were unpredictable and unexpected which lead to the discovery of the presently described technology.
  • Synthesis of l-homoarginine-d-amphetamine dihydrochloride (hArg-Amp) can be accomplished in three steps as shown in Scheme 3 below. In the first step, an N-protected hArg (e.g., Boc-hArg(NO2)) is coupled with d-amphetamine using EDCI. NHS is added to form an in-situ activated ester with DIPEA used as a co-base. The product is then subjected to hydrogenation under acidic conditions followed by deprotection with hydrochloric acid which forms the corresponding dihydrochloride salt.
    Figure imgb0005
  • The preparation of hArg-Amp required extensive modifications to previously published methods and syntheses. First, Boc-hArg(NO2)-OH required use of DMF to solubilize the material prior to reaction. Second, formation of the free base of amphetamine was performed in situ and was not isolated. Also, the formation of the activated ester was performed in situ with the addition reaction following in the same reaction vessel. Homoarginine is different from other standard and non-standard amino acids in that it requires a separate step of deprotection to remove the nitro group from the side chain. Failure to do so correctly can lead to undesirable products that do not perform in vivo with respect to the desired therapeutic outcomes discussed herein.
  • l-Citrulline-d-amphetamine hydrochloride (Cit-Amp) can be synthesized as shown in reaction Scheme 4 below in three overall steps. The first step involves the activation of Boc-Cit-OH to form an activated ester using DCC and NHS followed by the addition of d-amphetamine to produce the protected Boc-Cit-Amp. Deprotection using 4N HCl in dioxane gives the corresponding hydrochloride salt.
    Figure imgb0006
  • At least some compounds used in the present technology have no or a substantially decreased pharmacological activity when delivered through alternative routes of administration like intranasal or intravenous. However, they remain orally bioavailable at a level similar or slightly lower than other controlled release forms. The bioavailability can be a result of the hydrolysis of the covalent linkage following oral administration. Hydrolysis is time-dependent, thereby allowing amphetamine to become available in its active form over an extended period of time in a very controlled fashion. Therefore, the compounds used in the present technology can release amphetamine over an extended period and provide a therapeutically bioequivalent area under the curve (AUC) when compared to other controlled release forms of amphetamine (Adderall X® or Vyvanse™), with little or no spike in concentration max (Cmax) or equivalent Cmax. Not wanting to be bound by any particular theory, it is believed that since non-standard amino acids are used to produce the prodrug, the in vivo breakdown of the prodrug by enzymes would occur at a slower rate than, for example, when a standard amino acid is used to conjugate amphetamine. This will allow the prodrug to release amphetamine slowly and, preferably, only under in vivo conditions.
  • As a person of ordinary skill in the art will understand, drug products are considered pharmaceutical equivalents if they contain the same active ingredient(s), are of the same dosage form, route of administration and are identical in strength or concentration. Pharmaceutically equivalent drug products are formulated to contain the same amount of active ingredient in the same dosage form and to meet the same or compendial or other applicable standards (i.e., strength, quality, purity, and identity), but they may differ in characteristics such as shape, scoring configuration, release mechanisms, packaging, excipients (including colors, flavors, preservatives), expiration time, and, with certain limits, labeling. Drug products are considered to be therapeutic equivalents only if they are pharmaceutical equivalents and if they can be expected to have the same clinical effect and safety profile when administered to patients under the conditions specified in the labeling. The term "bioequivalent," on the other hand, describes pharmaceutical equivalent or pharmaceutical alternative products that display comparable bioavailability when studied under similar experimental conditions.
  • Standard amino acids such as lysine are not contemplated for the presently described technology, because lysine is an essential part of all dietary requirements, it would be expected that the prodrug conjugated with lysine would be released at a faster rate. By using non-standard amino acids, the release rate of amphetamine will be reduced due to the difference in overall digestion rate of non-standard amino acid conjugates of amphetamine versus standard amino acid conjugates of amphetamine such as lysine-amphetamine conjugate. This reduction in the rate of hydrolysis will decrease the incidence of cardiac side effects including higher blood pressure, rapid heart rate, and/or other subsequent side effects associated with conventional amphetamine treatment.
  • In accordance with the presently described technology, release of amphetamine after oral administration of the prodrug of the presently described technology would occur under desired physiological conditions. Preferably, other routes of administration (e.g., intranasal or intravenous) do not break the prodrug down to any appreciable extent. Also preferably, external means (chemical, enzymatic or other) will not break the prodrug down to any appreciable extent either. The breakdown ratio of the prodrug that can be achieved through external means is preferably less than about 50%, alternatively less than about 25%, alternatively less than about 20%, alternatively less than about 10%.
  • The presently described technology utilizes covalent modification of amphetamine by a non-standard amino acid to decrease its potential for causing behavioral deterioration or the rebound effect. It is believed that since the amphetamine is covalently modified to form the non-standard amino acid conjugate used in the present technology and releases slowly over the entire length of the day, little or no rebound effect can occur due to the slow continuous release of the active ingredient/drug/therapeutic component.
  • The presently described technology is also believed to provide reduced potential for rebound, reduced potential for abuse or addiction, and/or improve amphetamine's stimulant related toxicities. By limiting the blood level spike, doses are kept at levels required for a clinically significant effect without the unnecessary levels administered with other therapies. It is widely held that these spikes in blood levels can lead to cardiovascular toxicity in the form of higher blood pressure and rapid heart rate in addition to the euphoria encountered in drug abuse. Also, with a full day therapy, the risk of re-dosing is lowered, thus preventing additional toxicities or drug abuse issues.
  • The amphetamine prodrugs used in the presently described technology could be used for any condition requiring the stimulation of the central nervous system (CNS). These conditions include, for example, attention deficit hyperactivity disorder (ADHD), attention deficit disorder (ADD), obesity, narcolepsy, appetite suppressant, depression, anxiety, and wakefulness. Amphetamine stimulants have also demonstrated usefulness in treating stimulant (e.g., ***e, methamphetamine) abuse and addiction. Amphetamine stimulants have also been used extensively to improve battle field alertness and to combat fatigue.
  • Therefore, in accordance with some embodiments, the presently described technology provides amphetamine compositions comprising at least one amphetamine prodrug.
  • One embodiment is a composition that can prevent behavioral deterioration of amphetamine dosing comprising at least one ornithine conjugate of amphetamine.
  • Another embodiment is a composition for safely delivering amphetamine, comprising a therapeutically effective amount of at least one ornithine conjugate of amphetamine wherein the ornithine moiety can reduce the rate of absorption of the amphetamine as compared to delivering the unconjugated amphetamine or amphetamine conjugated to a standard amino acid.
  • Another embodiment of the present technology is a composition that can reduce amphetamine toxicity, comprising at least one ornithine conjugate of amphetamine wherein the ornithine moiety can release amphetamine over the entire course of a day providing a limited behavioral deterioration effect.
  • Another embodiment of the present technology is a composition that can reduce amphetamine toxicity, comprising at least one ornithine conjugate of amphetamine wherein the ornithine moiety can provide a serum release curve which does not increase above amphetamine's therapeutic level and does not cause blood level spiking.
  • Another embodiment of the present technology is a composition that can reduce bioavailability of amphetamine or prevent a toxic release profile in a patient, comprising at least one ornithine conjugate of amphetamine wherein the ornithine conjugate of amphetamine can maintain a steady-state serum release curve which can provide a therapeutically effective bioavailability but prevent spiking or increased blood serum concentrations compared to unconjugated amphetamine or amphetamine conjugated with a standard amino acid.
  • Another embodiment of the present technology is a composition comprising at least one ornithine conjugate of amphetamine that can prevent a Cmax or equivalent Cmax spike for amphetamine.
  • Another embodiment of the present technology is a composition comprising at least one ornithine conjugate of amphetamine that can prevent a Cmax or equivalent Cmax spike for amphetamine when taken by means other than orally while still providing a therapeutically effective bioavailability curve if taken orally.
  • In one or more embodiments, the ornithine conjugates of amphetamine used in the present technology may further comprise a polymer blend which comprises a hydrophilic polymer and/or a water-insoluble polymer. The polymers may be used according to industry standards to further enhance the sustained release/abuse resistant properties of the amphetamine prodrug used in the present technology without reducing the abuse resistance. For instance, a composition might include: about 70% to about 100% amphetamine prodrug by weight, from about 0.01% to about 10% of a hydrophilic polymer (e.g. hydroxypropyl methylcellulose), from about 0.01% to about 2.5% of a water-insoluble polymer (e.g. acrylic resin), from about 0.01% to about 1.5% of additives (e.g. magnesium stearate), and from about 0.01% to about 1% colorant by weight.
  • Hydrophilic polymers suitable for use in the sustained release formulations include one or more natural or partially or totally synthetic hydrophilic gums such as acacia, gum tragacanth, locust bean gum, guar gum, or karaya gum, modified cellulosic substances such as methylcellulose, hydroxymethylcellulose, hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethylcellulose, carboxymethylcellulose; proteinaceous substances such as agar, pectin, carrageen, and alginates; and other hydrophilic polymers such as carboxypolymethylene, gelatin, casein, zein, bentonite, magnesium aluminum silicate, polysaccharides, modified starch derivatives, and other hydrophilic polymers known to those of skill in the art, or a combination of such polymers. These hydrophilic polymers gel and would dissolve slowly in aqueous acidic media thereby allowing the amphetamine conjugate to diffuse from the gel in the stomach. When the gel reaches the intestines it would dissolve in controlled quantities in the higher pH medium to allow further sustained release. Preferred hydrophilic polymers are the hydroxypropyl methylcelluloses such as those manufactured by The Dow Chemical Company and known as Methocel ethers, such as Methocel E1OM.
  • Other formulations according to one or more embodiments of the present technology may further comprise pharmaceutical additives including, but not limited to, lubricants such as magnesium stearate, calcium stearate, zinc stearate, powdered stearic acid, hydrogenated vegetable oils, talc, polyethylene glycol, and mineral oil; colorants such as Emerald Green Lake, FD&C Red No. 40, FD&C Yellow No. 6, D&C Yellow No. 10, or FD&C Blue No. 1 and other various certified color additives (See 21 CFR, Part 74); binders such as sucrose, lactose, gelatin, starch paste, acacia, tragacanth, povidone polyethylene glycol, Pullulan and corn syrup; glidants such as colloidal silicon dioxide and talc; surface active agents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate, triethanolamine, polyoxyethylene sorbitan, poloxalkol, and quaternary ammonium salts; preservatives and stabilizers; excipients such as lactose, mannitol, glucose, fructose, xylose, galactose, sucrose, maltose, xylitol, sorbitol, chloride, sulfate and phosphate salts of potassium, sodium, and magnesium; and/or any other pharmaceutical additives known to those of skill in the art. In one preferred embodiment, a sustained release formulation of the present technology further comprises magnesium stearate and Emerald Green Lake.
  • The amphetamine compositions of the present technology, which comprises at least one amphetamine prodrug and can be further formulated with excipients, may be manufactured according to any appropriate method known to those of skill in the art of pharmaceutical manufacture. For instance, the amphetamine prodrug and a hydrophilic polymer may be mixed in a mixer with an aliquot of water to form a wet granulation. The granulation may be dried to obtain hydrophilic polymer encapsulated granules of the amphetamine prodrug. The resulting granulation may be milled, screened, then blended with various pharmaceutical additives such as, water insoluble polymers, and/or additional hydrophilic polymers. The formulation may then be tableted and may further be film coated with a protective coating which rapidly dissolves or disperses in gastric juices.
  • It should be noted that the above additives are not required for the amphetamine composition of the present technology to have sustained release in vivo properties. The non-standard amino acid conjugates used in the present technology are chemically stable to in vitro hydrolysis of the amide linkage to prevent tampering or removing the amphetamine prior to oral ingestion. Also, the controlled release of amphetamine through oral administration of the non-standard amino acid conjugate used in the present technology is an inherent property of the molecule, not related to the formulation. Put another way, the amphetamine prodrug used in the present technology itself can control the release of amphetamine into the digestive tract over an extended period of time resulting in an improved profile when compared to immediate release combinations and prevention of abuse without the addition of the above additives. Therefore, the prodrug used in the present technology can be easily formulated to different dosage forms. In one or more embodiments of the present technology, no further sustained release additives are required to achieve a blunted or reduced pharmacokinetic curve (e.g., reduced euphoric effect) while achieving therapeutically effective amounts of amphetamine release when taken orally.
  • The compositions of the presently described technology can be formulated into and administered by a variety of dosage forms through any oral routes of delivery. Once administered, the prodrugs will release amphetamine under digestive conditions. Any biologically-acceptable dosage form known to persons of ordinary skill in the art, and combinations thereof, are contemplated. Examples of preferred dosage forms include, without limitation, chewable tablets, quick dissolve tablets, effervescent tablets, reconstitutable powders, elixirs, liquids, solutions, suspensions, emulsions, tablets, multi-layer tablets, bi-layer tablets, capsules, soft gelatin capsules, hard gelatin capsules, caplets, lozenges, chewable lozenges, beads, powders, granules, particles, microparticles, dispersible granules, cachets, oral films (e.g., fast dissolving thin strips), and combinations thereof. Preferred dosage forms include capsule, solution formulation, and fast dissolving oral film.
  • Formulations of the present technology suitable for oral administration can be presented as discrete units, such as capsules, caplets, tablets, or oral films. These oral formulations also can comprise a solution or a suspension in an aqueous liquid or a non-aqueous liquid. The formulation can be an emulsion, such as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The oils can be administered by adding the purified and sterilized liquids to a prepared enteral formula, which can then be placed in the feeding tube of a patient who is unable to swallow.
  • If the capsule form is chosen, for example, excipients used in the capsule formulation could be broken up into four separate groups: bulk agent/binder, disintegrant, lubricant and carrier. A preferred capsule formulation comprises from about 50% to about 90% by weight a bulk agent such as various types of microcrystalline cellulose, from about 1% to about 5% by weight of a disintegrant such as croscarmellose sodium, from about 0.5% to about 2.5% of a lubricant such as magnesium state or other fatty acid salts. The carrier can be either hard gelatin capsules, and preferably use the smaller size ones such as #3 or #4 hard gelatin capsules.
  • Soft gel or soft gelatin capsules may be prepared, for example, by dispersing the formulation of the present technology in an appropriate vehicle (vegetable oils are commonly used) to form a high viscosity mixture. This mixture can then be encapsulated with a gelatin based film using technology and machinery known to those in the soft gel industry. The industrial units so formed are then dried to constant weight.
  • Chewable tablets, for example, may be prepared by mixing the formulations of the present technology with excipients designed to form a relatively soft, flavored, tablet dosage form that is intended to be chewed rather than swallowed. Conventional tablet machinery and procedures, that is both direct compression and granulation, i.e., or slugging, before compression, can be utilized. Those individuals involved in pharmaceutical solid dosage form production are versed in the processes and the machinery used as the chewable dosage form is a very common dosage form in the pharmaceutical industry.
  • Film-coated tablets, for example, may be prepared by coating tablets using techniques such as rotating pan coating methods or air suspension methods to deposit a contiguous film layer on a tablet.
  • Compressed tablets, for example, may be prepared by mixing the formulation of the present technology with excipients intended to add binding qualities to disintegration qualities. The mixture can be either directly compressed or granulated then compressed using methods and machinery known to those in the industry. The resultant compressed tablet dosage units are then packaged according to market need, i.e., unit dose, rolls, bulk bottles, blister packs, etc.
  • One preferred formulation is a fast dissolving oral film or thin strip. Methods and other ingredients needed to make oral films or thin strips are known in the art. Potential film forming agents include pullulan, hydroxypropylmethyl cellulose, hydroxypropyl cellulose, polyvinyl pyrrolidone, polyvinyl alcohol, sodium alginate, polyethylene glycol, xanthan gum, tragacanth gum, guar gum, acacia gum, Arabic gum, polyacrylic acid, amylase, starch, dextrin, pectin, chitin, chitosin, levan, elsinan, collagen, gelatin, zein, gluten, soy protein isolate, whey protein isolate, casein, and mixtures thereof.
  • Also, saliva stimulating agents, plasticizing agents, cooling agents, surfactants, emulsifying agents, thickening agents, binding agents sweeteners, flavoring, coloring agents, preservatives, or taste masking resins may be employed in the oral films or thin strips. Preferred agents include: pullulan, triethanol amine stearate, methyl cellulose, starch, triacetin, polysorbate 80, xanthan gum, maltitol, sorbitol and glycerol.
  • The presently described technology also contemplates the use of biologically-acceptable carriers which may be prepared from a wide range of materials. Without being limited thereto, such materials include diluents, binders and adhesives, lubricants, plasticizers, disintegrants, colorants, bulking substances, flavorings, sweeteners and miscellaneous materials such as buffers and adsorbents in order to prepare a particular medicated composition.
  • Binders may be selected from a wide range of materials such as hydroxypropylmethylcellulose, ethylcellulose, or other suitable cellulose derivatives, povidone, acrylic and methacrylic acid co-polymers, pharmaceutical glaze, gums, milk derivatives, such as whey, starches, and derivatives, as well as other conventional binders known to persons skilled in the art. Exemplary non-limiting solvents are water, ethanol, isopropyl alcohol, methylene chloride or mixtures and combinations thereof. Exemplary non-limiting bulking substances include sugar, lactose, gelatin, starch, and silicon dioxide.
  • Preferred plasticizers may be selected from the group consisting of diethyl phthalate, diethyl sebacate, triethyl citrate, cronotic acid, propylene glycol, butyl phthalate, dibutyl sebacate, castor oil and mixtures thereof, without limitation. As is evident, the plasticizers may be hydrophobic as well as hydrophilic in nature. Water-insoluble hydrophobic substances, such as diethyl phthalate, diethyl sebacate and castor oil are used to delay the release of water-soluble vitamins, such as vitamin B6 and vitamin C. In contrast, hydrophilic plasticizers are used when water-insoluble vitamins are employed which aid in dissolving the encapsulated film, making channels in the surface, which aid in nutritional composition release.
  • It should be understood that in addition to the ingredients particularly mentioned above, the formulations of the present technology can include other suitable agents such as flavoring agents, preservatives and antioxidants. Such antioxidants would be food acceptable and could include, for example, vitamin E, carotene, BHT or other antioxidants known to those of skill in the art.
  • Other compounds which may be included are, for example, medically inert ingredients, e.g., solid and liquid diluent, such as lactose, dextrose, saccharose, cellulose, starch or calcium phosphate for tablets or capsules, olive oil or ethyl oleate for soft capsules and water or vegetable oil for suspensions or emulsions; lubricating agents such as silica, talc, stearic acid, magnesium or calcium stearate and/or polyethylene glycols; gelling agents such as colloidal clays; thickening agents such as gum tragacanth or sodium alginate, binding agents such as starches, arabic gums, gelatin, methylcellulose, carboxymethylcellulose or polyvinylpyrrolidone; disintegrating agents such as starch, alginic acid, alginates or sodium starch glycolate; effervescing mixtures; dyestuff; sweeteners; wetting agents such as lecithin, polysorbates or laurylsulphates; and other therapeutically acceptable accessory ingredients, such as humectants, preservatives, buffers and antioxidants, which are known additives for such formulations.
  • For oral administration, fine powders or granules containing diluting, dispersing and/or surface-active agents may be presented in a draught, in water or a syrup, in capsules or sachets in the dry state, in a non-aqueous suspension wherein suspending agents may be included, or in a suspension in water or a syrup. Where desirable or necessary, flavoring, preserving, suspending, thickening or emulsifying agents can be included.
  • Liquid dispersions for oral administration may be syrups, emulsions or suspensions. The syrups may contain as carrier, for example, saccharose or saccharose with glycerol and/or mannitol and/or sorbitol. The suspensions and the emulsions may contain a carrier, for example a natural gum, agar, sodium alginate, pectin, methylcellulose, carboxymethylcellulose or polyvinyl alcohol.
  • The dose range for adult human beings will depend on a number of factors including the age, weight and condition of the patient. Suitable oral dosages of the prodrugs used in the presently described technology can be the equivalents of those typically found in amphetamine treatments. Typical dosages for amphetamine salts can range from about 1 mg to about 100 mg, although higher dosages may be approved at later dates. Using the molecular weight of the prodrug used in the present technology, the release percentage (% release) of amphetamine from the prodrug and desired dosage forms of the required amphetamine, the following equation can be generated: g r a m s o f a p r o d r u g n e e d e d = d o s a g e / m o l e c u l a r w e i g h t o f a m p h e t a m i n e % r e l e a s e m o l e c u l a r w e i g h t o f t h e p r o d r u g
    Figure imgb0007
  • Tablets, capsules, oral films, and other forms of presentation provided in discrete units conveniently contain a daily dose, or an appropriate fraction thereof, of one or more of the compounds used according to the invention. For example, units may contain from about 1 mg to about 500 mg, alternatively from about 5 mg to about 250 mg, alternatively from about 10 mg to about 100 mg of one or more of the compounds used in the presently described technology.
  • It is also possible for the dosage form of the present technology to combine any forms of release known to persons of ordinary skill in the art. These conventional release forms include immediate release, extended release, pulse release, variable release, controlled release, timed release, sustained release, delayed release, long acting, and combinations thereof. The ability to obtain immediate release, extended release, pulse release, variable release, controlled release, timed release, sustained release, delayed release, long acting characteristics and combinations thereof is known in the art.
  • Compositions of the present technology may be administered in a partial, i.e., fractional dose, one or more times during a 24 hour period, a single dose during a 24 hour period of time, a double dose during a 24 hour period of time, or more than a double dose during a 24 hour period of time. Fractional, double or other multiple doses may be taken simultaneously or at different times during the 24 hour period. The doses may be uneven doses with regard to one another or with regard to the individual components at different administration times.
  • Likewise, the compositions of the present technology may be provided in a blister pack or other such pharmaceutical package. Further, the compositions of the present technology may further include or be accompanied by indicia allowing individuals to identify the compositions as products for a prescribed treatment. The indicia may additionally include an indication of the above specified time periods for administering the compositions. For example, the indicia may be time indicia indicating a specific or general time of day for administration of the composition, or the indicia may be a day indicia indicating a day of the week for administration of the composition. The blister pack or other combination package may also include a second pharmaceutical product
  • It will be appreciated that the pharmacological activity of the compositions of the present technology can be demonstrated using standard pharmacological models that are known in the art. Furthermore, it will be appreciated that the compositions of the present technology can be incorporated or encapsulated in a suitable polymer matrix or membrane for site-specific delivery, or can be functionalized with specific targeting agents capable of effecting site specific delivery. These techniques, as well as other drug delivery techniques, are well known in the art.
  • In one or more embodiments of the present technology, the solubility and dissolution rate of the composition can be substantially changed under different physiological conditions encountered, for example, in the intestine, at mucosal surfaces, or in the bloodstream. In one or more embodiments of the present technology, the solubility and dissolution rate of the composition can substantially decrease the bioavailability of the amphetamine, particularly at doses above those intended for therapy. In one embodiment of the present technology, the decrease in bioavailability occurs upon intranasal administration. In another embodiment, the decrease in bioavailability occurs upon intravenous administration.
  • For each of the described embodiments of the present technology, one or more of the following characteristics can be realized: The cardiovascular toxicity of the amphetamine prodrug is substantially lower than that of the unconjugated amphetamine and amphetamine conjugated with a standard amino acid. The covalently bound non-standard amino acid moiety reduces or eliminates the possibility of behavioral deterioration or the rebound effect. The covalently bound non-standard amino acid moiety reduces or eliminates the possibility of abuse by intranasal administration. The covalently bound non-standard amino acid moiety reduces the possibility of abuse by injection.
  • The presently described technology provides the possibility for altering and/or delivering amphetamines in a manner that can decrease their potential for abuse. The present technology provides various ways to regulate pharmaceutical dosage through conjugating amphetamine with non-standard amino acids.
  • In one embodiment behavioral deterioration or the rebound effect is prevented by administering to a patient in need an amphetamine prodrug composition of the present technology.
  • In another embodiment the possibility for safely delivering amphetamine is provided wherein a therapeutically effective amount of at least one ornithine conjugate of amphetamine is provided wherein the ornithine moiety can reduce the rate of absorption of amphetamine as compared to delivering the unconjugated amphetamine or amphetamine conjugated with a standard amino acid.
  • In another embodiment the possibility for reducing amphetamine cardiovascular toxicity is provided wherein a patient is provided with at least one ornithine conjugate of amphetamine, wherein the ornithine moiety can decrease the rate of release of amphetamine within the first a few hours of administration.
  • In another embodiment the possibility for reducing amphetamine cardiovascular toxicity is provided wherein a patient is provided with at least one ornithine conjugate of amphetamine, wherein the ornithine moiety can provide a serum release curve which does not increase above the amphetamine's cardiovascular toxicity level.
  • In another embodiment the possibility for reducing bioavailability of amphetamine or for preventing a toxic release profile of amphetamine in a patient, is provided wherein at least one ornithine conjugate of amphetamine is provided, wherein the conjugated amphetamine can maintain a steady-state serum release curve which provides a therapeutically effective bioavailability but prevents spiking or increased blood serum concentrations compared to amphetamine conjugated with a standard amino acid.
  • In another embodiment the possibility for preventing a Cmax or equivalent Cmax spike for amphetamine is provided while still providing a therapeutically effective bioavailability curve wherein at least one ornithine conjugate of amphetamine, is administered to a patient.
  • In another embodiment of the present technology the possibility for reducing or preventing abuse of amphetamine is provided wherein a composition is provided, administered, consumed, on prescribed to a patient in need thereof, wherein said composition comprises at least one ornithine conjugate of amphetamine such that the pharmacological activity of amphetamine is decreased when the composition is used in a manner inconsistent with the manufacturer's instructions.
  • In another embodiment of the present technology the possibility of preventing behavioral deterioration or the rebound effect of amphetamine or stimulant treatment is provided wherein an amphetamine composition of the presently described technology is provided administered, consumed, or prescribed to a patient in need thereof, wherein said composition comprises at least one ornithine conjugate of amphetamine that can decrease the potential of behavioral deterioration or the rebound effect from amphetamine or stimulant treatment.
  • In another embodiment of the present technology the possibility for reducing or preventing the euphoric effect of amphetamine is provided wherein a composition comprising at least one ornithine conjugate of amphetamine that can decrease the pharmacological activity of amphetamine when the composition is used in a manner inconsistent with the manufacturer's instructions, is provided, administered, consumed or provided to a human in need thereof.
  • In any of the preceding possibilities the amphetamine composition used may be adapted for oral administration, and the amphetamine prodrug may be resistant to release amphetamine from the ornithine moiety when the composition is administered parenterally, such as intranasally or intravenously. Preferably, amphetamine may be released from the ornithine moiety in the presence of the intestinal tract. Optionally, the amphetamine composition used may be in the form of a tablet, capsule, oral film, oral solution, oral suspension, or other oral dosage form discussed herein.
  • The composition of the present technology may yield a therapeutic effect without substantial euphoria. Preferably, the amphetamine composition of the present technology can provide a therapeutically bioequivalent AUC when compared to other controlled release amphetamine compositions but does not provide a Cmax which results in euphoria or an equivalent Cmax.
  • The following properties may be achieved through conjugating amphetamine to the non-standard amino acid ornithine. In one embodiment, the cardiovascular toxicity or stress of the non-standard amino acid conjugate of amphetamine used in the present technology may be lower than that of the amphetamine when the amphetamine is delivered in its unconjugated state, as a compound conjugated to a standard amino acid, or as a salt thereof. In another embodiment, the possibility of behavioral deterioration is reduced or eliminated. In another embodiment, the possibility of abuse by intranasal administration is reduced or eliminated. In another embodiment, the possibility of abuse by intravenous administration is reduced or eliminated.
  • The compositions of the present technology may be used in methods of treating various diseases or conditions requiring the stimulation of the central nervous system (CNS) comprising administering compositions of the present technology which, optionally, further comprise commonly prescribed active agents for the respective illness or disease. For instance, in one embodiment of the invention) the compositions of the present technology may be used in a method of treating attention deficit hyperactivity disorder (ADHD)
    or treating attention deficit disorder (ADD).
  • In another embodiment of the invention, the compositions of the present technology may be used in a method of treating narcolepsy comprising administering to a patient compositions of the presently described technology.
  • The presently described technology and its advantages will be better understood by reference to the following examples. These examples are provided to describe specific embodiments of the present technology.
  • Example 1: Comparative study of pharmacokinetic parameters of released d-amphetamine following administration of a non-standard amino acid conjugate (hArg-Amp) and a standard amino acid conjugate (Vyvanse™, Lys-Amp)
  • The pharmacokinetic parameters of d-amphetamine following oral administration of a non-standard amino acid conjugate and a standard amino acid conjugate, Vyvanse™ (Lys-Amp), commercially available from Shire, Incorporated of Wayne, PA are studied in this example. The non-standard amino acid conjugate used in this example is the hydrochloride salt of hArg-Amp. The results are recorded in the table below: Table 1
    Parameter Non-standard amino acid % amp1 Vyvanse™ % total Amp2
    AUC 0-8h 94% 100%
    AUC0-4h 77% 100%
    AUCinf 95% 100%
    Cmax 76% 100%
    T
    max 400% 100%
    1Percent amphetamine released relative to Vyvanse™ (at an equimolar concentration of amphetamine contained in the non-standard amino acid prodrug compared to the total amphetamine contained in Vyvanse™)
    2 Percent amphetamine relative to 50mg Vyvanse™ dose
  • The study shows that the Cmax of hArg-Amp of is significantly lower than that of Vyvanse™, a standard amino acid conjugate of d-amphetamine, which can lead to lower cardiovascular effects (blood pressure, heart rate). Quick release (higher Cmax) of amphetamine has already demonstrated significant increases in blood pressure and heart rate. In certain patient populations, these cardiovascular side effects can be dose limiting or can cause the termination of stimulant therapy.
  • The pharmacokinetic parameters of d-amphetamine following parental administration of hArg-Amp and d-amphetamine are also studied. The study shows that little release of amphetamine (<50%) happens when hArg-Amp is taken through parental routes (intranasal, intravenous) due to differences in enzymes encountered in gut versus other routes. When Adderall X® or other controlled release formulations of amphetamine are injected or snorted, the pharmacokinetic parameters of the amphetamine are significantly altered and an individual can use these changes to produce euphoria.
  • Example 2: Preparation of Boc-Orn(Boc)-Amp
  • Boc-Orn(Boc)-OH (1.5 g, 4.518 mmol) was dissolved in DMF (15 ml). EDCI (1.299 g, 6.777 mmol), NHS (0.572 g, 4.969 mmol), d-apmphetamine (0.732 g, 5.422 mmol) and DIEA (0.87 ml, 4.969 mmol) were then added sequentially. The clear reaction mixture was stirred at room temperature for 16 hours (hrs). The reaction mixture was quenched with pH 3 water (40 ml), and the product was extracted with EtOAc (3 x 70 ml). The combined extracts were washed with pH 3 water, saturated NaHCO3 followed by water. The EtOAc layer was dried over anhydrous Na2SO4. Solvent was removed to obtain 1.82 g of protected amide as a white solid.
  • The white solid was analyzed by 1H NMR (CDCl3) δ. The results show 1.1-1.2 (m, 3H, Amp α-CH3), 1.3-1.5 (m, 18H, Boc CH3), 1.6-1.8 (m, 4H, Orn β, γ CH2), 2.75 (m, 2H, Amp β CH2), 3.05-3.1 (m, 2H, Orn δ CH2), 3.2 (m, 1H, Amp α CH), 4.1 (m, 1H, Orn α CH), 7.1-7.4 (m, 5H, Amp Ar-H). These NMR shifts are consistant with the structure of Orn-Amp.
  • Example 3: Preparation of Orn-Amp
  • Boc-Orn(Boc)-Amp (1.35 g, 3 mmol) was dissolved in EtOAc (200 ml) and to the slightly cloudy solution was added MsOH (0.43 ml, 6.6 mmol) drop wise. The reaction mixture became a clear solution which was stirred at room temperature for approximately 20 hrs. Solvent was removed and the residue was triturated in hexanes. Off-white solid product was formed which was filtered under vacuum and washed with hexanes. The solid was dried in vacuum oven for 20 hrs to obtain 0.88 g of Orn-Amp·2MsOH (1-ornithine-d-amphetamine dimesylate).
  • The product obtained was tested by 1H NMR (DMSO-d6) δ. The result shows 1.1 (m, 3H, Amp α-CH3), 1.4-1.6 (m, 4H, Orn β, γ CH2), 2.35 (s, 6H, CH3SO3H CH3), 2.6-2.8 (m, 4H, Amp β and Orn δ), 3.75 (m, 1H, Amp α), 4.05 (m, 1H, Orn α), 7.1-7.3 (m, 5H, Amp Ar-H), 7.6-8.5 (br peaks, amide and amine); 13C NMR (DMSO-d6) δ 18.45 (Om γ), 21.49 (Orn β), 27.30 (Amp β), 37.38 (Amp CH3), 37.77 (Amp α), 41.20 (Om δ), 51.54 (Orn α), 125.29 (p-Ar), 127.27 (m-Ar), 129.17 (o-Ar), 137 (Ar), 166.58 (C=O); M+1 = 250.7. These results are consistent with the proposed structure.
  • Example 4: Reparation of Boc-hAre(NO 2 )-Amp
  • Boc-hArg(NO2)-OH (2.667 g, 8 mmol) was dissolved in DMF (25 ml). EDCI (2.30 g, 12 mmol), NHS (1.012 g, 8.8 mmol), d-amphetamine (1.269 g, 9.6 mmol) and DIEA (1.138 g, 8.8 mmol) were then added sequentially. The clear reaction mixture was stirred at room temperature for 16 hrs. The reaction mixture was quenched with pH 3 water (150 ml), and the product was extracted with EtOAc (3 x 50 ml). The combined extracts were washed with pH 3 water followed by saturated NaCl. The EtOAc layer was dried over anhydrous MgSO4. The product was recrystallized from EtOAc-Hexane two times to give 2.36 g of desired protected product.
  • The product was analyzed using 1H NMR (DMSO-d6) 8. The result shows 0.9-1.1 (m, 3H, Amp CH3), 1.1-1.2 (m, 2H, hArg γ CH2), 1.2-1.5 (m, 13H, Boc CH3, hArg β,δ CH2), 2.55-2.75 (m, 2H, Amp β CH2), 3.1 (m, 2H, hArg ε CH2), 3.75 (m, 1H, Amp α CH), 3.95 (m, 1H, hArg α CH), 6.65 (t, 1H, hArg guanidino NH), 7.1-7.3 (m, 5H, Amp Ar-H), 7.6-8.2 (br m, 2H, hArg guanidine NH and amide NH), 8.5 (br s, 1H, hArg NH-NO2). These results would be considered consistent with the proposed structure.
  • Example 5: Preparation of hArg-Amp-2HCl (1-homoarginine-d-amphetamine dihydrochloride)
  • Boc-hArg(NO2)-Amp (1.5 g) was dissolved in HPLC grade MeOH (120 ml) and to the clear solution was added the Pd-C catalyst (10%, Aldrich). A small stir bar was placed in the flask and the reaction mixture was stirred under a slow stream of hydrogen overnight after incorporating the 5-6N HCl in 2-propanol solution (1.5ml). After the overnight reaction, the solution was filtered and the solvent evaporated. The white crystalline product was dried under vacuum to give 1.61 g of the Boc-hArg-Amp intermediate product.
  • The product (1.6 g) was dissolved in 80 ml of HPLC grade MeOH, and 5-6N HCl in 2-propanol (3.2 mL) was added to the solution. The reaction mixture was stirred overnight, solvent removed and re-dissolved in minimum amount of MeOH. The final product was crashed out with MTBE, and dried under vacuum at 30 °C for about 20 hours to yield 1.12 g of a white powder.
  • The white powder was analyzed using 1H NMR (DMSO-d6) δ. The result shows 0.9-1.1 (m, 3H, Amp CH3), 1.1-1.2 (m, 2H, hArg γ CH2), 1.35 (m, 2H, hArg β CH2), 1.55(mn, 2H, hArg δ CH2), 2.75 (d, 2H, Amp β CH2), 3.0 (m, 2H, hArg ε CH2), 3.75 (m, 1H, Amp α CH), 4.05 (m, 1H, hArg α CH), 7.1-7.2 (m, 5H, Amp Ar-H), 7.2-7.8 (br m, 3H, amide NH, HCl), 8.0 (t, 1H, hArg guanidino NH), 8.2 (br s, 2H, amide or guanidino NH2), 8.75 (d, 1H, amide NH); 13C NMR (DMSO-d6) δ 21.08 (Amp CH3), 21.36 (hArg γ), 28.23 (hArg δ), 32.28 (hArg β), 40.18 (Amp β), 42.19 (hArg ε), 46.88 (Amp α), 52.23 (hArg α), 126.54 (p-Ar), 128.52 (m-Ar), 129.60 (o-Ar), 139.34 (Ar), 157.61 (C=O), 167.95 (guanidino C); M+1 = 306. These results would be considered to be consistant with the proposed structure.
  • Example 6: Preparation of Cit-Amp·HCl (l-citrulline-d-amphetamine hydrochloride)
  • Boc-Cit-OH (0.500 g, 1.82 mmol) was dissolved in anhydrous THF. To this solution was added NHS (0.209 g, 1.82 mmol) followed by DCC (0.376 g, 1.82 mmol). Resulting slurry was stirred at ambient temperature overnight. In a separate flask, d-amphetamine sulfate (0.306 g, 0.83 mmol) was suspended in THF (10 ml) and NMM (0.34 ml, 3.64 mmol) was added. The activated ester was filtered directly into the amphetamine suspension and the resulting suspension was stirred overnight. The reaction was quenched with 5% NaHCO3 and IPAC for 45 min. Organic solvent was then removed. The aqueous layer was then extracted 3 times with IPAC and the combined organics were washed with 5% acetic acid, 5% NaHCO3 and 5% NaCl. The organic layer was then dried over NaSO4 and solvent was removed. Crude product was recrystallized using IPAC/heptane to yield 200 mg of a white solid.
  • 10 ml of 4N HCl in dioxane were added to the 200 mg (0.200 g) Boc-Cit-Amp. The mixture was stirred at room temperature for 6 hours and solvent was removed.
  • Example 7: Comparative biological study of Lys-Amp and hArg-Amp
  • Male Sprague-Dawley rats were fasted overnight and dosed by oral gavage with either l-homoarginine-d-amphetamine (hArg-Amp) or l-lysine-d-amphetamine (Vyvanse™, Lys-Amp). Water was provided ad libitum. Doses were calculated at an equivalent 1.5mg/mg freebase equivalent of d-amphetamine. Plasma concentrations of d-amphetamine were measured using ELISA (Neogen Corp. Lexington, KY).
  • Mean plasma concentration curves (n=5) of d-amphetamine released by l-homoarginine-d-amphetamine or l-lysine-d-amphetamine are shown in Figure 1. Pharmacokinetic(PK) parameters of this study are listed in Table 2. Table 2. Pharmacokinetic Properties of hArg-Amp and Lys-Amp
    Vehicle % AUC Tmax Cmax % Tmax % Cmax
    Lys-Amp 100% 3h 44 ng/ml 100% 100%
    hArg-Amp 99% 4h 44 ng/ml 133% 100%
  • This pharmacokinetic (PK) study clearly demonstrates a shift in the Tmax for the non-standard amino acid (hArg-Amp) compared to the standard amino acid (Lys-Amp). This shift may be due to a reduction in the rate of enzymatic hydrolysis of the amide bond of the non-standard amino acid attached to amphetamine vs. the standard amino acid attached to amphetamine.
  • Figures 2-4 represent different ways to view the data reflected in Figure 1 and Table 2. As further discussed below, these figures highlight the differences of hArg-Amp over Lys-Amp during the first several hours.
  • Figure 2 demonstrates the relative blood levels of d-amphetamine released from both Lys-Amp and hArg-Amp. The graph shows that equivalent blood levels do not occur until later time points and that blood levels do not appear to spike or have a more significant Cmax than Lys-Amp. The amount of d-amphetamine released from hArg-Amp is gradual and maintains a more steady concentration over the duration of the study than did Lys-Amp. In contrast, Lys-Amp blood levels of released d-amphetamine "spiked" at 3 hours and cleared more quickly than the blood levels obtained from hArg-Amp.
  • Figures 3 and 4 show the difference in blood levels obtained from the study described in Figure 2. As is shown, the initial blood levels for both conjugates (Lys-Amp and hArg-Amp) are very different, with hArg-Amp releasing amphetamine at a more gradual rate. These differences in blood levels become less during the more critical duration of action for stimulant treatments and more importantly, the differences are greater again at later time points suggesting that hArg-Amp maintains a more consistent dose of amphetamine when compared to Lys-Amp. The longer duration of release for hArg-Amp would suggest a much lower opportunity for behavioral deterioration to occur.
  • Other oral studies have been conducted in a similar fashion and are summarized in Table 3 below. The average PK results for four (4) oral studies (n=20 per vehicle) are recorded in Figure 5: Table 3. Average Results of 4 Oral Studies (n=20 per compound)
    Vehicle % AUC Tmax % Tmax % Cmax % AUC 0-4h
    Lys-Amp 100% 1h 100% 100% 100%
    hArg-Amp 94% 4h 400% 76% 77%
  • Example 8: Biological study of hArg-Amp and Orn-Amp
  • To compare the amount of release of d-amphetamine among various non-standard amino acids, l-ornithine-d-amphetamine (Orn-Amp) was dosed in replace of Lys-Amp in Example 7 in another oral pharmacokinetic study. Mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp or Orn-Amp are shown in Figure 6. Pharmacokinetic parameters of this study are listed in Table 4. Table 4. Pharmacokinetic Properties of hArg-Amp and Orn-Amp
    Vehicle % AUC Tmax Cmax % Tmax % Cmax
    hArg-Amp 100% 4h 32 ng/ml 100% 100%
    Orn-Amp 78% 1h 27 ng/ml 25% 84%
  • Example 9: Comparative biological study of Lys-Amp, hArg-Amp, Orn-Amp and Cit-Amp
  • To compare the amount of release of d-amphetamine among various non-standard amino acids, l-ornithine-d-amphetamine (Orn-Amp), hArg-Amp and l-citrulline-d-amphetamine (Cit-Amp) were dosed with Lys-Amp in another oral pharmacokinetic study. Mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp, Orn-Amp and Cit-Amp are shown in Figure 7. Pharmacokinetic parameters of this study are listed in Table 5.
  • Direct comparison of 3 non-standard amino acid conjugates of amphetamine (Cit, Orn and hArg) demonstrate the significant ability to shift or change the pharmacokinetic properties versus the standard amino acids. All non-standard amino acids studied released amphetamine in an amount greater than 50%. Ornithine and homoarginine both showed Cmax levels far below that of lysine and both homoarginine and citrulline significantly shifted the Tmax compared to Lys-Amp. These changes to the pharmacokinetic properties of amphetamine when conjugated to non-standard amino acids represent clinically significant changes not described or demonstrated by Lys-Amp nor described or demonstrated by other standard amino acids. Table 5: Oral Properties of Lys-Amp, hArg-Amp, Orn-Amp and Cit-Amp
    Vehicle % AUC Tmax Cmax % Tmax % Cmax
    Lys-Amp 100% 1h 59 ng/ml 100% 100%
    hArg-Amp 68% 2h 27 ng/ml 200% 46%
    Orn-Amp 52% 1h 32 ng/ml 100% 544%
    Cit-Amp 95% 15m 129 ng/ml 25% 219%
  • Example 10: Intranasal study of Amp, hArg-Amp, Orn-Amp
  • Male Sprague-Dawley rats were fasted overnight and dosed by intranasal administration with either hArg-Amp, Orn-Amp or d-amphetamine. Doses were calculated at an equivalent 1.5mg/kg freebase equivalent of d-amphetamine. Plasma concentrations of d-amphetamine were measured using ELISA. Mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp or Orn-Amp are shown in Figure 8. Pharmacokinetic parameters of this study are listed in Table 6. No significant release (<25%) was observed in either hArg-Amp or Orn-Amp. Table 6. Intranasal Properties of d-Amp, hArg-Amp and Orn-Amp
    Vehicle % AUC Tmax Cmax % Tmax % Cmax
    d-amp 100% 15m 53 ng/ml 100% 100%
    hArg-Amp 23% 2h 9 ng/ml 1600% 17%
    Orn-Amp 14% 2h 10 ng/ml 1600% 19%
  • Example 11: Intravenous study of Amp, hArg-Amp, Orn-Amp
  • Male Sprague-Dawley rats were dosed by intravenous administration through the tail vein with hArg-Amp, Orn-Amp or d-amphetamine. Doses were calculated at an equivalent 1.5mg/kg freebase equivalent of d-amphetamine. Plasma concentrations of d-amphetamine were measured using ELISA. Mean plasma concentration curves (n=5) of d-amphetamine released by hArg-Amp or Orn-Amp are shown in Figure 9. Pharmacokinetic parameters of this study are listed in Table 7. No significant release (<35%) was observed in either hArg-Amp or Orn-Amp. The initial spike in d-amphetamine released from hArg-Amp cleared quickly while as in the intranasal study, Orn-Amp had a slight increase at the 2 hour point. Table 7. Intravenous Properties of d-Amp, hArg-Amp and Orn-Amp
    Vehicle % AUC Tmax Cmax % Tmax % Cmax
    d-amp 100% 15m 396 ng/ml 100% 100%
    hArg-Amp 41% 15m 135 ng/ml 100% 34%
    Orn-Amp 10% 15m 26 ng/ml 100% 7%
  • Results of the studies in Examples 7-11 clearly show an unexpected change in the oral pharmacokinetic properties by using non-standard amino acids over standard amino acids. By changing the non-standard amino acid attached to amphetamine, the conjugates are able to shift Tmax (earlier or later), modify curve shape, lower Cmax, and raise Cmax. In addition, the shift in Tmax for hArg-Amp may be clinically significant in that many of the cardiovascular side effects and toxicity are related to Tmax and Cmax. The results demonstrate that by using non-standard amino acids a shift in the Tmax, with a lower Cmax occurs without changing AUC significantly. In addition, the slope of uptake of hArg-Amp vs. Lys-Amp appears to be more gradual thus leading to a slower onset which could further alleviate side effects.
  • The amphetamine conjugate, hArg-Amp, demonstrates that by using non-standard amino acids, a shift in the Tmax occurs while still retaining AUC and potential clinical effect. By using non-standard amino acids, we are able to demonstrate that both hArg-Amp and Orn-Amp show little release via the IN and IV route yet still maintain a similar AUC.
  • The presently described technology is now described in such full, clear, concise and exact terms as to enable any person skilled in the art to which it pertains, to practice the same. It is to be understood that the foregoing describes preferred embodiments of the invention.

Claims (16)

  1. A composition for treating a human or animal patient having a disorder or condition requiring the stimulation of the central nervous system of the patient, comprising at least one conjugate of amphetamine with ornithine, a salt thereof, a derivative thereof, or a combination thereof.
  2. The composition of claim 1, wherein at least one conjugate of amphetamine is l-ornithine-d-amphetamine.
  3. The composition of claim 1, wherein the composition has a reduced or prevented pharmacological activity when administered by parenteral routes.
  4. The composition of claim 1, wherein the salt of the conjugate of amphetamine is a mesylate, a hydrochloride salt, a sulfate, an oxalate, a triflate, a citrate, a malate, a tartrate, a phosphate, a nitrate, a benzoate, or a mixture thereof.
  5. The composition of claim 1, wherein the composition is in the form of a tablet, a capsule, a caplet, a troche, a lozenge, an oral powder, a solution, an oral film, a thin strip, or a suspension.
  6. The composition of claim 5, wherein the tablet, troche, or lozenge is chewable.
  7. The composition of claim 1, wherein at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof is present in the amount of from about 1 mg to about 500 mg, preferably from about 5 mg to about 250 mg, more preferably from about 10 mg to about 100 mg.
  8. The composition of claim 1, wherein at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof is in an amount sufficient to provide a therapeutically bioequivalent AUC when compared to amphetamine alone, but does not provide a Cmax spike or does not provide an equivalent Cmax.
  9. Use of at least one conjugate of amphetamine with ornithine, a salt thereof, a derivative thereof, or a combination thereof for the preparation of a pharmaceutical composition for treating a patient having a disorder or condition requiring the stimulation of the central nervous system of the patient, wherein the pharmaceutical composition is to be administered orally to the patient.
  10. The use of claim 9, wherein at least one conjugate of amphetamine is l-ornithine-d-amphetamine.
  11. The use of claim 9, wherein said oral administration comprises administering a tablet, a capsule, a caplet, a troche, a lozenge, an oral powder, an oral solution, an oral film, a thin strip, or an oral suspension.
  12. The use of claim 9, wherein the pharmaceutical composition is for administering the conjugate of amphetamine in the form of a salt.
  13. The use of claim 12, wherein the salt is a mesylate, a hydrochloride salt, a sulfate, an oxalate, a triflate, a citrate, a malate, a tartrate, a phosphate, a nitrate, a benzoate, or a mixture thereof.
  14. The use of claim 9, wherein the pharmaceutical composition contains from about 1 mg to about 500 mg, preferably from about 5 mg to about 250 mg, more preferably from about 10 mg to about 100 mg of the at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof.
  15. The use of claim 9, wherein the pharmaceutical composition is for administering at least one conjugate of amphetamine, the salt thereof, the derivative thereof, or the combination thereof in an amount sufficient to provide a therapeutically bioequivalent AUC when compared to amphetamine alone, but does not provide a Cmax spike or does not provide an equivalent Cmax.
  16. The use of claim 9, wherein the disorder or condition is attention deficit hyperactivity disorder, attention deficit disorder, obesity, narcolepsy, appetite suppression, depression, anxiety, wakefulness, or a combination thereof.
EP07869098A 2006-12-11 2007-12-10 Ornithine conjugates of amphetamine and processes for making and using the same Not-in-force EP2101571B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US86937506P 2006-12-11 2006-12-11
PCT/US2007/087028 WO2008073918A1 (en) 2006-12-11 2007-12-10 Non-standard amino acid conjugates of amphetamine and processes for making and using the same

Publications (3)

Publication Number Publication Date
EP2101571A1 EP2101571A1 (en) 2009-09-23
EP2101571A4 EP2101571A4 (en) 2010-01-13
EP2101571B1 true EP2101571B1 (en) 2011-05-18

Family

ID=39512105

Family Applications (1)

Application Number Title Priority Date Filing Date
EP07869098A Not-in-force EP2101571B1 (en) 2006-12-11 2007-12-10 Ornithine conjugates of amphetamine and processes for making and using the same

Country Status (12)

Country Link
US (3) US7776917B2 (en)
EP (1) EP2101571B1 (en)
JP (1) JP2010512413A (en)
CN (1) CN101594778A (en)
AT (1) ATE509622T1 (en)
AU (1) AU2007333105A1 (en)
BR (1) BRPI0720105A2 (en)
CA (1) CA2672138A1 (en)
ES (1) ES2364865T3 (en)
HK (1) HK1131518A1 (en)
IL (1) IL198930A0 (en)
WO (1) WO2008073918A1 (en)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060014697A1 (en) * 2001-08-22 2006-01-19 Travis Mickle Pharmaceutical compositions for prevention of overdose or abuse
CA2645855C (en) 2006-03-16 2015-02-03 Tris Pharma, Inc. Modified release formulations containing drug-ion exchange resin complexes
ATE509622T1 (en) 2006-12-11 2011-06-15 Kempharm Inc AMPHETAMINE-ORNITHINE CONJUGATES AND PROCESS OF PRODUCTION AND USE
BRPI0807157A2 (en) * 2007-02-08 2014-04-29 Kempharm Inc POLY HYDROPHYAL PROPHARMACIES OF AMPHTAMINE AND OTHER STIMULANTS AND PROCESSES FOR THE MANUFACTURE AND USE OF THE SAME
US8614346B2 (en) 2009-06-19 2013-12-24 Cambrex Charles City, Inc. Methods and compositions for preparation of amphetamine conjugates and salts thereof
WO2011133348A1 (en) 2010-04-21 2011-10-27 Pharmacofore, Inc. Compositions comprising enzyme-cleavable amphetamine prodrugs and inhibitors thereof
US8779191B2 (en) 2010-12-20 2014-07-15 Cambrex Charles City, Inc. Methods and compositions for preparing lisdexamfetamine and salts thereof
EP3189840B1 (en) * 2011-07-28 2021-05-19 Kempharm, Inc. Methylphenidate-prodrugs, processes of making and using the same
EP2739140A4 (en) * 2011-07-29 2015-06-03 Shire Llc Homoarginine prodrugs and/or conjugates of amphetamine and other stimulants and processes for making and using the same
EP2872161B1 (en) 2012-06-26 2020-12-16 Del Mar Pharmaceuticals Dianhydrogalactitol for use in treating tyrosine-kinase-inhibitor-resistant malignancies in patients with genetic polymorphisms or ahi1 dysregulations or mutations
US20140073589A1 (en) * 2012-06-27 2014-03-13 Shire Ag Amphetamine Prodrugs
WO2014058742A1 (en) * 2012-10-09 2014-04-17 Sears Douglas Therapeutic treatment
US10314839B2 (en) 2014-10-20 2019-06-11 Elysium Therapeutics, Inc. Diversion-resistant opioid formulations
US20170056352A1 (en) 2015-08-25 2017-03-02 Rgenix, Inc. PHARMACEUTICALLY ACCEPTABLE SALTS OF beta-GUANIDINOPROPIONIC ACID WITH IMPROVED PROPERTIES AND USES THEREOF
US11590228B1 (en) 2015-09-08 2023-02-28 Tris Pharma, Inc Extended release amphetamine compositions
US10335406B2 (en) 2015-10-01 2019-07-02 Elysium Therapeutics, Inc. Opioid compositions resistant to overdose and abuse
EP3355885A4 (en) 2015-10-01 2019-04-24 Elysium Therapeutics, Inc. Polysubunit opioid prodrugs resistant to overdose and abuse
KR102499414B1 (en) 2016-11-15 2023-02-13 클라리아 파마 홀딩 에이비 pharmaceutical preparation
KR102337138B1 (en) 2016-12-11 2021-12-08 켐팜 인코포레이티드 Compositions comprising methylphenidate-prodrugs, methods of making and using same
WO2018160178A1 (en) 2017-03-01 2018-09-07 Rgenix, Inc. Pharmaceutically acceptable salts of b-guanidinopropionic acid with improved properties and uses thereof
JP7234130B2 (en) 2017-03-17 2023-03-07 エリージウム セラピューティクス, インコーポレイテッド Polysubunit opioid prodrugs resistant to overdose and abuse
CA3059781A1 (en) * 2017-04-10 2018-10-18 Shire Pharmaceuticals Inc. Methods of treatment using an amphetamine prodrug
GB201709141D0 (en) 2017-06-08 2017-07-26 Klaria Pharma Holding Ab Pharmaceutical formulation
US11590081B1 (en) 2017-09-24 2023-02-28 Tris Pharma, Inc Extended release amphetamine tablets
GB2571696B (en) 2017-10-09 2020-05-27 Compass Pathways Ltd Large scale method for the preparation of Psilocybin and formulations of Psilocybin so produced
WO2019199731A1 (en) * 2018-04-10 2019-10-17 Shire Pharmaceuticals Inc. Amphetamine prodrug and crystalline froms thereof
GB201808462D0 (en) * 2018-05-23 2018-07-11 Klaria Pharma Holding Ab Pharmaceutical formulation
TW202103699A (en) 2019-04-17 2021-02-01 英商康派斯百事威公司 Treatment of depression and other various disorders with psilocybin

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE539185A (en) * 1954-06-28
US2881113A (en) * 1957-01-29 1959-04-07 Ortho Pharma Corp Therapeutically active compositions containing amphetamines
US2892753A (en) * 1957-02-26 1959-06-30 Boehringer Sohn Ingelheim Central nervous system stimulant
DE1493824A1 (en) 1964-01-27 1969-05-22 Hoffmann La Roche Process for the preparation of aminocarboxylic acid amides
CZ299283B6 (en) 1997-12-22 2008-06-04 Euro-Celtique, S. A. Medicinal form for peroral administration
WO2001013921A1 (en) 1999-08-23 2001-03-01 Ockert David M Triple drug therapy for the treatment of narcotic and alcohol withdrawal symptoms
CA2411564A1 (en) 2000-06-09 2001-12-13 The Regents Of The University Of California Method of treating pain using nalbuphine and opioid antagonists
US6417184B1 (en) 2000-09-19 2002-07-09 David M. Ockert Triple drug therapy for the treatment and prevention of acute or chronic pain
KR100822498B1 (en) 2002-02-22 2008-04-16 샤이어 엘엘씨 Novel Sustained Release Pharmaceutical Compounds to Prevent Abuse of Controlled Substances
US7105486B2 (en) * 2002-02-22 2006-09-12 New River Pharmaceuticals Inc. Abuse-resistant amphetamine compounds
US7691603B2 (en) 2003-04-09 2010-04-06 Novo Nordisk A/S Intracellular formation of peptide conjugates
SI1644019T2 (en) 2003-05-29 2018-04-30 Shire Llc Abuse resistant amphetamine compounds
EP1931357A2 (en) 2005-09-13 2008-06-18 Shire LLC Prodrugs of phentermine
ATE509622T1 (en) 2006-12-11 2011-06-15 Kempharm Inc AMPHETAMINE-ORNITHINE CONJUGATES AND PROCESS OF PRODUCTION AND USE
BRPI0807157A2 (en) 2007-02-08 2014-04-29 Kempharm Inc POLY HYDROPHYAL PROPHARMACIES OF AMPHTAMINE AND OTHER STIMULANTS AND PROCESSES FOR THE MANUFACTURE AND USE OF THE SAME

Also Published As

Publication number Publication date
US20090234018A1 (en) 2009-09-17
US20080139653A1 (en) 2008-06-12
BRPI0720105A2 (en) 2014-10-07
EP2101571A1 (en) 2009-09-23
ES2364865T3 (en) 2011-09-15
US20100292336A1 (en) 2010-11-18
HK1131518A1 (en) 2010-01-29
US7776917B2 (en) 2010-08-17
US8101661B2 (en) 2012-01-24
CA2672138A1 (en) 2008-06-19
CN101594778A (en) 2009-12-02
ATE509622T1 (en) 2011-06-15
WO2008073918A1 (en) 2008-06-19
JP2010512413A (en) 2010-04-22
IL198930A0 (en) 2010-02-17
EP2101571A4 (en) 2010-01-13
AU2007333105A1 (en) 2008-06-19

Similar Documents

Publication Publication Date Title
EP2101571B1 (en) Ornithine conjugates of amphetamine and processes for making and using the same
EP2109465B1 (en) Polar hydrophilic prodrugs of amphetamine and other stimulants and processes for making and using the same
KR101159477B1 (en) Abuse resistant amphetamine compounds
US20140171510A1 (en) Homoarginine prodrugs and/or conjugates of amphetamine and other stimulants and processes for making and using the same
US20220064116A1 (en) D-amphetamine compounds, compositions, and processes for making and using the same

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090609

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC MT NL PL PT RO SE SI SK TR

A4 Supplementary search report drawn up and despatched

Effective date: 20091210

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 47/48 20060101ALI20091205BHEP

Ipc: A61K 31/198 20060101ALI20091205BHEP

Ipc: A61K 31/135 20060101AFI20091205BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1131518

Country of ref document: HK

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20100222

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/198 20060101ALI20101122BHEP

Ipc: A61K 31/135 20060101AFI20101122BHEP

Ipc: A61K 47/48 20060101ALI20101122BHEP

RTI1 Title (correction)

Free format text: ORNITHINE CONJUGATES OF AMPHETAMINE AND PROCESSES FOR MAKING AND USING THE SAME

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602007014748

Country of ref document: DE

Effective date: 20110630

REG Reference to a national code

Ref country code: NL

Ref legal event code: VDEP

Effective date: 20110518

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2364865

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20110915

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1131518

Country of ref document: HK

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110919

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110918

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110819

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: KEMPHARM, INC.

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20120221

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20120111

Year of fee payment: 5

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20111228

Year of fee payment: 5

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602007014748

Country of ref document: DE

Effective date: 20120221

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20120103

Year of fee payment: 5

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20111231

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20111210

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20111231

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20111231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20111210

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110818

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20120127

Year of fee payment: 5

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20121210

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20130830

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110518

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130702

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602007014748

Country of ref document: DE

Effective date: 20130702

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130102

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20121210

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20121210

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20140306

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20121211